Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Hepatol ; 70(6): 1170-1179, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30654068

RESUMO

BACKGROUND & AIMS: Inherited abnormalities in apolipoprotein E (ApoE) or low-density lipoprotein receptor (LDLR) function result in early onset cardiovascular disease and death. Currently, the only curative therapy available is liver transplantation. Hepatocyte transplantation is a potential alternative; however, physiological levels of hepatocyte engraftment and repopulation require transplanted cells to have a competitive proliferative advantage of over host hepatocytes. Herein, we aimed to test the efficacy and safety of a novel preparative regimen for hepatocyte transplantation. METHODS: Herein, we used an ApoE-deficient mouse model to test the efficacy of a new regimen for hepatocyte transplantation. We used image-guided external-beam hepatic irradiation targeting the median and right lobes of the liver to enhance cell transplant engraftment. This was combined with administration of the hepatic mitogen GC-1, a thyroid hormone receptor-ß agonist mimetic, which was used to promote repopulation. RESULTS: The non-invasive preparative regimen of hepatic irradiation and GC-1 was well-tolerated in ApoE-/- mice. This regimen led to robust liver repopulation by transplanted hepatocytes, which was associated with significant reductions in serum cholesterol levels after transplantation. Additionally, in mice receiving this regimen, ApoE was detected in the circulation 4 weeks after treatment and did not induce an immunological response. Importantly, the normalization of serum cholesterol prevented the formation of atherosclerotic plaques in this model. CONCLUSIONS: Significant hepatic repopulation and the cure of dyslipidemia in this model, using a novel and well-tolerated preparative regimen, demonstrate the clinical potential of applying this method to the treatment of inherited metabolic diseases of the liver. LAY SUMMARY: Hepatocyte transplantation is a promising alternative to liver transplantation for the treatment of liver diseases. However, it is inefficient, as restricted growth of transplanted cells in the liver limits its therapeutic benefits. Preparative treatments improve the efficiency of this procedure, but no clinically-feasible options are currently available. In this study we develop a novel well-tolerated preparative treatment to improve growth of cells in the liver and then demonstrate that this treatment completely cures an inherited lipid disorder in a mouse model.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Dislipidemias/terapia , Hepatócitos/transplante , Hiperlipoproteinemia Tipo II/terapia , Acetatos/farmacologia , Animais , Apolipoproteínas E/sangue , Colesterol/sangue , Modelos Animais de Doenças , Feminino , Hepatócitos/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenóis/farmacologia
2.
Hepatol Commun ; 7(3): e0070, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36848094

RESUMO

BACKGROUND: α1-antitrypsin deficiency is most commonly caused by a mutation in exon-7 of SERPINA1 (SA1-ATZ), resulting in hepatocellular accumulation of a misfolded variant (ATZ). Human SA1-ATZ-transgenic (PiZ) mice exhibit hepatocellular ATZ accumulation and liver fibrosis. We hypothesized that disrupting the SA1-ATZ transgene in PiZ mice by in vivo genome editing would confer a proliferative advantage to the genome-edited hepatocytes, enabling them to repopulate the liver. METHODS: To create a targeted DNA break in exon-7 of the SA1-ATZ transgene, we generated 2 recombinant adeno-associated viruses (rAAV) expressing a zinc-finger nuclease pair (rAAV-ZFN), and another rAAV for gene correction by targeted insertion (rAAV-TI). PiZ mice were injected i.v. with rAAV-TI alone or the rAAV-ZFNs at a low (7.5×1010vg/mouse, LD) or a high dose (1.5×1011vg/mouse, HD), with or without rAAV-TI. Two weeks and 6 months after treatment, livers were harvested for molecular, histological, and biochemical analyses. RESULTS: Two weeks after treatment, deep sequencing of the hepatic SA1-ATZ transgene pool showed 6%±3% or 15%±4% nonhomologous end joining in mice receiving LD or HD rAAV-ZFN, respectively, which increased to 36%±12% and 36%±12%, respectively, 6 months after treatment. Two weeks postinjection of rAAV-TI with LD or HD of rAAV-ZFN, repair by targeted insertion occurred in 0.10%±0.09% and 0.25%±0.14% of SA1-ATZ transgenes, respectively, which increased to 5.2%±5.0% and 33%±13%, respectively, 6 months after treatment. Six months after rAAV-ZFN administration, there was a marked clearance of ATZ globules from hepatocytes, and resolution of liver fibrosis, along with reduction of hepatic TAZ/WWTR1, hedgehog ligands, Gli2, a TIMP, and collagen content. CONCLUSIONS: ZFN-mediated SA1-ATZ transgene disruption provides a proliferative advantage to ATZ-depleted hepatocytes, enabling them to repopulate the liver and reverse hepatic fibrosis.


Assuntos
Edição de Genes , Nucleases de Dedos de Zinco , Humanos , Animais , Camundongos , Cirrose Hepática/genética , Cirrose Hepática/terapia , Hepatócitos , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular
3.
Hepatology ; 50(3): 815-24, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19585550

RESUMO

UNLABELLED: Asialoglycoprotein receptor (ASGPR)-mediated endocytosis has been used to target genes to hepatocytes in vivo. However, the level and duration of transgene expression have been low because of lysosomal translocation and degradation of the DNA and lack of its integration into the host genome. In this study we packaged the DNA of interest in proteoliposomes containing the fusogenic galactose-terminated F-glycoprotein of the Sendai virus (FPL) for targeted delivery to hepatocytes. After the FPL binds to ASGPR on the hepatocyte surface, fusogenic activity of the F-protein delivers the DNA into the cytosol, bypassing the endosomal pathway. For transgene integration we designed plasmids containing one transcription unit expressing the Sleeping Beauty transposase (SB) and another expressing human uridinediphosphoglucuronate glucuronosyltransferase-1A1 (pSB-hUGT1A1). The latter was flanked by inverted/direct repeats that are substrates of SB. In cell culture, FPL-mediated delivery of the E. coli beta-galactosidase gene (LacZ) resulted in transduction of ASGPR-positive cells (rat hepatocytes or Hepa1 cell line), but not of ASGPR-negative 293 cells. Intravenous injection of the FPL-entrapped pSB-hUGT1A1 (4-8 microg/day, 1-4 doses) into UGT1A1-deficient hyperbilirubinemic Gunn rats (model of Crigler-Najjar syndrome type 1) resulted in hUGT1A1 expression in 5%-10% of hepatocytes, but not in other cell types. Serum bilirubin levels declined by 30% +/- 4% in 2 weeks and remained at that level throughout the 7-month study duration. With histidine containing FPL, serum bilirubin was reduced by 40% +/- 5%, and bilirubin glucuronides were excreted into bile. No antibodies were detectable in the recipient rats against the F-protein or human UGT1A1. CONCLUSION: FPL is an efficient hepatocyte-targeted gene delivery platform in vivo that warrants further exploration toward clinical application.


Assuntos
Receptor de Asialoglicoproteína/administração & dosagem , Icterícia/terapia , Proteolipídeos/administração & dosagem , Transposases/administração & dosagem , Animais , Síndrome de Crigler-Najjar/terapia , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Terapia Genética/métodos , Glucuronosiltransferase/administração & dosagem , Hepatócitos/efeitos dos fármacos , Humanos , Hiperbilirrubinemia/terapia , Ratos , Ratos Gunn , Proteínas Virais de Fusão/administração & dosagem
4.
Hepatology ; 49(1): 258-67, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19003915

RESUMO

Engraftment of donor hepatocytes is a critical step that determines the success of hepatocyte transplantation. Rapid and efficient integration of donor cells would enable prompt liver repopulation of these cells in response to selective proliferative stimuli offered by a preparative regimen. We have earlier demonstrated that hepatic irradiation (HIR) in combination with a variety of hepatotrophic growth signals, such as partial hepatectomy and hepatocyte growth factor, can be used as a preparative regimen for liver repopulation of transplanted hepatocytes. In this study, we investigated the effects of HIR on engraftment of transplanted dipeptidyl peptidase IV (DPPIV)-positive hepatocytes in congeneic DPPIV-deficient rats. HIR-induced apoptosis of hepatic sinusoidal endothelial cells (SEC) within 6 hours of HIR resulted in dehiscence of the SEC lining in 24 hours. Although there was no change of the number of Kupffer cells after HIR, colloidal carbon clearance decreased 24 hours post HIR, indicating a suppression of phagocytic function. DPPIV+ donor cells were transplanted 24 hours after HIR (0-50 Gy). There was an HIR dose-dependent increase in the donor hepatocyte mass engrafted in the liver parenchyma. The number of viable transplanted hepatocytes present in hepatic sinusoids or integrated in the parenchyma was greater in the HIR-treated group at 3 and 7 days after transplantation compared with the sham controls. Finally, we validated these rodent studies in cynomolgus monkeys, demonstrating that a single 10-Gy dose of HIR was sufficient to enhance engraftment of donor porcine hepatocytes. These data indicate that transient disruption of the SEC barrier and inhibition of the phagocytic function of Kupffer cells by HIR enhances hepatocyte engraftment and the integrated donor cell mass. Thus, preparative HIR could be potentially useful to augment hepatocyte transplantation.


Assuntos
Hepatócitos/efeitos da radiação , Hepatócitos/transplante , Fígado/cirurgia , Animais , Apoptose , Proliferação de Células/efeitos da radiação , Dipeptidil Peptidase 4/genética , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Feminino , Sobrevivência de Enxerto , Hepatectomia , Células de Kupffer/fisiologia , Células de Kupffer/efeitos da radiação , Fígado/citologia , Regeneração Hepática/fisiologia , Macaca fascicularis , Masculino , Fagocitose/efeitos da radiação , Ratos , Ratos Endogâmicos F344 , Suínos , Condicionamento Pré-Transplante/métodos , Transplante Heterólogo
5.
Dev Cell ; 4(5): 607-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12737794

RESUMO

Two nuclear receptors of xenobiotic drugs, PXR and CAR, are central regulators of detoxification enzymes. New studies extend the role of these receptors to a natural detoxification process. They coordinate induction of proteins for storage, glucuronidation, and canalicular transport of bilirubin.


Assuntos
Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Fatores de Transcrição/metabolismo , Animais , Bilirrubina/metabolismo , Receptor Constitutivo de Androstano , Glucuronosiltransferase/metabolismo , Humanos , Inativação Metabólica , Receptor de Pregnano X
6.
J Pediatr ; 154(4): 616-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19324225

RESUMO

A 6-day-old female newborn, readmitted for extreme hyperbilirubinemia with bilirubin encephalopathy, died despite 2 double-volume exchange transfusions. On autopsy examination the basal ganglia and hippocampus were selectively stained deep yellow. The infant was heterozygous for both the glucose-6-phosphate dehydrogenase Mediterranean mutation and for the (TA)(6)/(TA)(7) promoter polymorphism for the gene encoding the bilirubin conjugating enzyme uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1*28, associated with Gilbert syndrome). No additional mutations of the UGT1A1 were detected. Seemingly innocuous, heterozygotic mutations may interact synergistically to result in serious and even fatal outcomes.


Assuntos
Deficiência de Glucosefosfato Desidrogenase/complicações , Kernicterus/etiologia , Evolução Fatal , Feminino , Glucosefosfato Desidrogenase/genética , Deficiência de Glucosefosfato Desidrogenase/genética , Glucosiltransferases/genética , Heterozigoto , Humanos , Recém-Nascido , Judeus/genética , Kernicterus/patologia , Polimorfismo Genético
7.
Methods Mol Biol ; 481: 117-40, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19096805

RESUMO

Ex vivo gene transfer into hepatocytes could serve several purposes in the context of gene therapy or cell transplantation: (1) isolated hepatocytes can be transduced in culture with therapeutic genes and then transplanted into the recipient; (2) marker genes can be introduced for subsequent identification of transplanted cells and their progeny; (3) gene transfer can be used for conditional immortalization of hepatocytes for expansion in culture; (4) immunomodulatory genes can be transferred into hepatocytes to prevent allograft rejection. Gene transfer into cultured hepatocytes can be achieved using DNA that is not incorporated into recombinant viruses. In such systems, transgene integration into the host cell genome can be enhanced using transposon systems, such as "sleeping beauty." In addition to using the conventional reagents, such as cationic liposomes, DNA transfer into hepatocytes can be achieved by Nucleofection or special hepatocyte-targeted carriers such as proteoliposomes containing galactose-terminated glycoproteins (e.g. the F protein of the Sendai virus). Alternatively, genes can be transferred using recombinant viruses, such as adenoviral vectors that are episomal or retroviral vectors (including lentiviruses) that permit integration of the transgene into the host genome. Gene transfer using lentiviral vectors has been achieved in both attached and suspended hepatocytes. Transduction efficiency of lentiviral vectors can be enhanced using magnetic nanoparticles (Magnetofection).


Assuntos
Técnicas de Transferência de Genes , Hepatócitos/metabolismo , Animais , Linhagem Celular Transformada/citologia , Terapia Genética/métodos , Hepatócitos/citologia , Hepatócitos/transplante , Humanos , Transplante de Fígado/métodos , Magnetismo/métodos , Modelos Biológicos , Nanopartículas/uso terapêutico , Coloração e Rotulagem/métodos , Condicionamento Pré-Transplante/métodos
8.
Int J Radiat Oncol Biol Phys ; 103(4): 922-934, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30503786

RESUMO

Orthotopic liver transplantation (OLT) has been effective in managing end-stage liver disease since the advent of cyclosporine immunosuppression therapy in 1980. The major limitations of OLT are organ supply, monetary cost, and the burden of lifelong immunosuppression. Hepatocyte transplantation, as a substitute for OLT, has been an exciting topic of investigation for several decades. HT is potentially minimally invasive and can serve as a vehicle for delivery of personalized medicine through autologous cell transplant after modification ex vivo. However, 3 major hurdles have prevented large-scale clinical application: (1) availability of transplantable cells; (2) safe and efficient ex vivo gene therapy methods; and (3) engraftment and repopulation efficiency. This review will discuss new sources for transplantable liver cells obtained by lineage reprogramming, clinically acceptable methods of genetic manipulation, and the development of hepatic irradiation-based preparative regimens for enhancing engraftment and repopulation of transplanted hepatocytes. We will also review the results of the first 3 patients with genetic liver disorders who underwent preparative hepatic irradiation before hepatocyte transplantation.


Assuntos
Hepatócitos/citologia , Transplante de Fígado/métodos , Animais , Proliferação de Células , Terapia Genética , Humanos , Transplante de Fígado/efeitos adversos , Segurança
9.
Hepatol Commun ; 3(1): 129-146, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30620000

RESUMO

Hepatocyte transplantation is an attractive alternative to liver transplantation. Thus far, however, extensive liver repopulation by adult hepatocytes has required ongoing genetic, physical, or chemical injury to host liver. We hypothesized that providing a regulated proliferative and/or survival advantage to transplanted hepatocytes should enable repopulation in a normal liver microenvironment. Here, we repopulated livers of DPPIV- (dipeptidyl peptidase-4) rats and Ugt1a1 (uridinediphosphoglucuronate glucuronosyltransferase 1a1)-deficient Gunn rats (model of Crigler-Najjar syndrome type 1), both models without underlying liver injury, for up to 1 year by transplanting lenti-hYAP-ERT2 (mutated estrogen receptor ligand-binding domain 2)-transduced hepatocytes (YAP-Hc). Yap (yes-associated protein) nuclear translocation/function in YAP-Hc was regulated by tamoxifen. Repopulating YAP-Hc and host hepatocytes were fluorescence-activated cell sorting-purified and their transcriptomic profiles compared by RNAseq. After 1 year of liver repopulation, YAP-Hc clusters exhibited normal morphology, integration into hepatic plates and hepatocyte-specific gene expression, without dysplasia, dedifferentiation, or tumorigenesis. RNAseq analysis showed up-regulation of 145 genes promoting cell proliferation and 305 genes suppressing apoptosis, including hepatocyte growth factor and connective tissue growth factor among the top 30 in each category and provided insight into the mechanism of cell competition that enabled replacement of host hepatocytes by YAP-Hc. In Gunn rats transplanted with YAP-Hc+tamoxifen, there was a 65%-81% decline in serum bilirubin over 6 months versus 8%-20% with control-Hc, representing a 3-4-fold increase in therapeutic response. This correlated with liver repopulation as demonstrated by the presence of Ugt1a1-positive hepatocyte clusters in livers and western blot analysis of tissue homogenates. Conclusion: Tamoxifen-regulated nuclear translocation/function of hYAP-ERT2 enabled long-term repopulation of DPPIV-/- and Gunn rat livers by hYAP-ERT2-transduced hepatocytes without tumorigenesis. This cell transplantation strategy may offer a potential therapy for most of the inherited monogenic liver diseases that do not exhibit liver injury.

10.
Transplantation ; 85(9): 1253-60, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18475180

RESUMO

BACKGROUND: Primary hyperoxaluria type-1 (PH1) is an autosomal recessive disease characterized by excessive oxalate production by hepatocytes caused by the deficiency of peroxisomal alanine-glyoxylate aminotransferase (AGT) activity. Persistent hyperoxaluria causes nephrocalcinosis and urolithiasis, leading to renal failure, followed by tissue oxalosis with life-threatening complications. Combined liver-kidney transplantation is the only definitive treatment of PH1. Hepatocyte transplantation, which is much less invasive, could have offered an attractive alternative. However, because the AGT-deficient hepatocytes overproduce oxalate, a large fraction of the mutant host hepatocytes must be replaced by AGT-competent cells, which is beyond the capacity of current hepatocyte transplantation procedures. Here, we have evaluated a preparative irradiation-based method of liver repopulation in an Agxt-deleted mouse model of PH1 (Agxt-/-). MATERIALS AND METHODS: Hepatocytes (10(6) viable cells) isolated from congeneic mice ([ROSA]26 C57BL/6J) expressing Escherichia coli beta-galactosidase were transplanted into Agxt-/- mice by intrasplenic injection. The preparative regimen consisted of X-irradiation of the host liver and mitotic stimulation of the hepatocytes by adenovector-based expression of hepatocyte growth factor. RESULTS: The procedure resulted in progressive replacement of the mutant host hepatocytes with the AGT-competent hepatocytes, leading to correction of urinary oxalate excretion. Oral ethylene glycol challenge (0.7% for 1 week) resulted in nephrocalcinosis and microlithiasis in untreated Agxt-/- mice, but not in the mice after hepatic repopulation. CONCLUSION: The results indicate that hepatocyte transplantation after appropriate preparative regimens may permit sufficient repopulation of the liver to ameliorate hyperoxaluria, and therefore should be evaluated further as a potential treatment of PH1.


Assuntos
Hepatócitos/transplante , Hiperoxalúria Primária/cirurgia , Animais , Modelos Animais de Doenças , Etilenoglicol/administração & dosagem , Hepatócitos/enzimologia , Regeneração Hepática , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrocalcinose/etiologia , Transaminases/deficiência , Transplante Homólogo , beta-Galactosidase/metabolismo
11.
Clin Pharmacol Drug Dev ; 6(2): 140-146, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28263463

RESUMO

Hyperbilirubinemia is a common finding in individuals with a history of substance abuse. Although this may indicate a serious disorder of liver function, this is not always the case. An understanding of bilirubin formation, metabolism, and transport can provide a helpful approach to dealing with these patients. This is typified by studies of patients treated with the antiretroviral drug atazanavir. Atazanavir has been associated with hyperbilirubinemia in as many as one-third of individuals for whom it has been prescribed, evoking concerns of hepatotoxicity. The studies in this report were designed to determine mechanisms by which this occurs. The data show that this drug inhibits the enzyme UDP-glucuronosyl transferase-1A1, responsible for conjugating bilirubin with glucuronic acid. This conjugation step is required for bilirubin excretion into bile, and when it is inhibited, bilirubin refluxes from the liver into the circulation, causing unconjugated hyperbilirubinemia. Other parameters of bilirubin formation, binding to albumin in the circulation, uptake into hepatocytes, and intracellular protein binding in hepatocytes were unaffected by atazanavir. The effect of atazanavir on serum bilirubin levels is reversible, consistent with lack of structural damage to the liver.


Assuntos
Sulfato de Atazanavir/efeitos adversos , Bilirrubina/sangue , Inibidores da Protease de HIV/efeitos adversos , Hiperbilirrubinemia/induzido quimicamente , Animais , Sulfato de Atazanavir/administração & dosagem , Bilirrubina/metabolismo , Células Cultivadas , Feminino , Glucuronosiltransferase/antagonistas & inibidores , Glutationa Transferase/metabolismo , Inibidores da Protease de HIV/administração & dosagem , Hepatócitos/metabolismo , Humanos , Hiperbilirrubinemia/sangue , Masculino , Ratos Wistar , Ritonavir/administração & dosagem , Transtornos Relacionados ao Uso de Substâncias/complicações
12.
Hepatol Int ; 11(1): 54-69, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27530815

RESUMO

The discovery that coordinated expression of a limited number of genes can reprogram differentiated somatic cells to induced pluripotent stem cells (iPSC) has opened novel possibilities for developing cell-based models of diseases and regenerative medicine utilizing cell reprogramming or cell transplantation. Directed differentiation of iPSCs can potentially generate differentiated cells belonging to any germ layer, including cells with hepatocyte-like morphology and function. Such cells, termed iHeps, can be derived by sequential cell signaling using available information on embryological development or by forced expression of hepatocyte-enriched transcription factors. In addition to the translational aspects of iHeps, the experimental findings have provided insights into the mechanisms of cell plasticity that permit one cell type to transition to another. However, iHeps generated by current methods do not fully exhibit all characteristics of mature hepatocytes, highlighting the need for additional research in this area. Here we summarize the current approaches and achievements in this field and discuss some existing hurdles and emerging approaches for improving iPSC differentiation, as well as maintaining such cells in culture for increasing their utility in disease modeling and drug development.


Assuntos
Técnicas de Cultura de Células/métodos , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Humanos , Medicina Regenerativa , Transdução de Sinais
13.
Methods Mol Biol ; 1506: 131-147, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27830550

RESUMO

Liver transplantation has been established as a curative therapy for acute and chronic liver failure, as well as liver-based inherited metabolic diseases. Because of the complexity of organ transplantation and the worldwide shortage of donor organs, hepatocyte transplantation is being developed as a bridging therapy until donor organs become available, or for amelioration of inherited liver-based diseases. The Gunn rat is a molecular and metabolic model of Crigler-Najjar syndrome type 1, which is characterized by lifelong unconjugated hyperbilirubinemia due to the lack of uridinediphosphoglucuronate glucuronosyltransferase-1 (UGT1A1)-mediated bilirubin glucuronidation. Gunn rats are convenient for evaluating the effect of hepatocyte transplantation or gene therapy, because the extent of UGT1A1 replacement can be assessed by serial determination of serum bilirubin levels, and excretion of bilirubin glucuronides in bile provide definitive evidence of the function of the transplanted hepatocytes or the effect of gene therapy. The core techniques involved in hepatocyte transplantation in Gunn rats are discussed in this chapter.


Assuntos
Transplante de Células/métodos , Síndrome de Crigler-Najjar/cirurgia , Técnicas de Transferência de Genes , Hepatócitos/transplante , Hepatopatias/cirurgia , Animais , Bile/química , Pigmentos Biliares/análise , Bilirrubina/análogos & derivados , Bilirrubina/sangue , Bilirrubina/metabolismo , Separação Celular/instrumentação , Separação Celular/métodos , Cromatografia Líquida de Alta Pressão , Síndrome de Crigler-Najjar/sangue , Modelos Animais de Doenças , Feminino , Terapia Genética/métodos , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Hepatócitos/metabolismo , Heterozigoto , Homozigoto , Humanos , Hiperbilirrubinemia/sangue , Fígado/metabolismo , Fígado/cirurgia , Hepatopatias/metabolismo , Testes de Função Hepática , Masculino , Ratos , Ratos Gunn
14.
Biochem J ; 392(Pt 3): 685-92, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16171463

RESUMO

Bilirubin glucuronidation, catalysed by UGT1A1 [UGT (UDP glucuronosyltransferase) isoform 1A1, EC 2.4.1.17], is critical for biliary elimination of bilirubin. UGT1A1 deficiency causes CN-1 (Crigler-Najjar syndrome type 1), which is characterized by potentially lethal unconjugated hyperbilirubinaemia. Nucleotide sequence analysis of UGT1A1 in two CN-1 patients revealed that patient A was homozygous for a nt 530 G-->A (where nt 530 G-->A means guanine to adenine transition at nucleotide 530) mutation, predicting a C177Y substitution, and patient B had a nt 466 T-->C mutation on one allele and a nt 1070 A-->G mutation on the other, predicting a C156R and a Q357R substitution respectively. All 11 cysteine residues of mature human UGT1A1 are highly conserved in other human UGT isoforms and in rat, mouse and Rhesus monkey UGT1A1, suggesting their functional importance. Expression of mutagenized UGT1A1 plasmids showed that substitution of any of the seven cysteine residues located within the endoplasmic reticulum cisternae (including those mutated in patients A and B) abolished UGT1A1 activity or markedly increased its apparent K(m) for bilirubin. Substitution of the three cysteine residues within the C-terminal cytosolic tail had minimal effect on basal UGT1A1 activity, but prevented UGT1A1 activation by UDP-GlcNAc. N-Ethylmaleimide did not inhibit UGT1A1 activity in native microsomes, but prevented UGT1A1 activation by UDP-GlcNAc and inhibited the activity in digitonin-permeabilized microsomes. Dithiothreitol did not affect UGT1A1 activity in human liver microsomes. Together, the results suggested that free thiol groups, but not disulphide bonding, of seven cysteine residues within the intracisternal region of human UGT1A1 are important for its catalytic activity, while cysteine residues in the cytosolic domain may be involved in its physiological activation by UDP-GlcNAc.


Assuntos
Cisteína/metabolismo , Glucuronosiltransferase/química , Glucuronosiltransferase/metabolismo , Sequência de Aminoácidos , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Glucuronosiltransferase/genética , Humanos , Lactente , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Dados de Sequência Molecular , Mutação Puntual , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
15.
Cell Transplant ; 25(12): 2221-2243, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27512979

RESUMO

Although several types of somatic cells have been reprogrammed into induced pluripotent stem cells (iPSCs) and then differentiated to hepatocyte-like cells (iHeps), the method for generating such cells from renal tubular epithelial cells shed in human urine and transplanting them into animal livers has not been described systematically. We report reprogramming of human urinary epithelial cells into iPSCs and subsequent hepatic differentiation, followed by a detailed characterization of the newly generated iHeps. The epithelial cells were reprogrammed into iPSCs by delivering the pluripotency factors OCT3/4, SOX2, KLF4, and MYC using methods that do not involve transgene integration, such as nucleofection of episomal (oriP/EBNA-1) plasmids or infection with recombinant Sendai viruses. After characterization of stable iPSC lines, a three-step differentiation toward hepatocytes was performed. The iHeps expressed a large number of hepatocyte-preferred genes, including nuclear receptors that regulate genes involved in cholesterol homeostasis, bile acid transport, and detoxification. MicroRNA profile of the iHeps largely paralleled that of primary human hepatocytes. The iHeps engrafted into the livers of Scid mice transgenic for mutant human SERPINA1 after intrasplenic injection. Thus, urine is a readily available source for generating human iHeps that could be potentially useful for disease modeling, pharmacological development, and regenerative medicine.


Assuntos
Células Epiteliais/citologia , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Células Epiteliais/metabolismo , Citometria de Fluxo , Hepatócitos/metabolismo , Humanos , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariotipagem , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Urotélio/citologia
16.
Stem Cell Reports ; 5(1): 22-30, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26074313

RESUMO

Hepatocyte transplantation has the potential to cure inherited liver diseases, but its application is impeded by a scarcity of donor livers. Therefore, we explored whether transplantation of hepatocyte-like cells (iHeps) differentiated from human induced pluripotent stem cells (iPSCs) could ameliorate inherited liver diseases. iPSCs reprogrammed from human skin fibroblasts were differentiated to iHeps, which were transplanted into livers of uridinediphosphoglucuronate glucuronosyltransferase-1 (UGT1A1)-deficient Gunn rats, a model of Crigler-Najjar syndrome 1 (CN1), where elevated unconjugated bilirubin causes brain injury and death. To promote iHep proliferation, 30% of the recipient liver was X-irradiated before transplantation, and hepatocyte growth factor was expressed. After transplantation, UGT1A1+ iHep clusters constituted 2.5%-7.5% of the preconditioned liver lobe. A decline of serum bilirubin by 30%-60% and biliary excretion of bilirubin glucuronides indicated that transplanted iHeps expressed UGT1A1 activity, a postnatal function of hepatocytes. Therefore, iHeps warrant further exploration as a renewable source of hepatocytes for treating inherited liver diseases.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Síndrome de Crigler-Najjar/terapia , Hepatócitos/transplante , Hiperbilirrubinemia/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Animais , Bilirrubina/sangue , Síndrome de Crigler-Najjar/sangue , Síndrome de Crigler-Najjar/patologia , Glucuronosiltransferase/deficiência , Glucuronosiltransferase/genética , Humanos , Hiperbilirrubinemia/sangue , Hiperbilirrubinemia/genética , Fígado/patologia , Fígado/cirurgia , Ratos , Ratos Gunn
17.
BioDrugs ; 17(2): 81-91, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12641487

RESUMO

Current treatment modalities available for hepatitis B virus (HBV) or hepatitis C virus (HCV) infections are not efficient. The enormous disease burden caused by these two infections makes the development of novel therapies critical. For HCV, the development of an effective vaccine is urgent in view of the escalating number of infected individuals. Molecular therapies for HBV and HCV infection can be directed at reducing viral load by interfering with the life cycle of the viruses or at generating immune response against viral epitopes. The antiviral approaches consist of the delivery or expression of antisense RNAs, ribozymes or dominant negative proteins. Viral biology can be interrupted by attacking various potential targets within the two viruses. DNA-based vaccination strategies are being explored for both prevention and treatment of these diseases. Both non-viral and recombinant viral vectors are being developed for safe, effective and long-term gene transfer to the liver. Although no "ideal" vector is available at this time, the ingenuity of numerous investigators is leading to the improvement of the vector systems, promising successful application of gene therapy to the prevention and treatment of viral hepatitis in the foreseeable future.


Assuntos
Hepatite B/terapia , Hepatite C/terapia , Animais , Antivirais/uso terapêutico , Terapia Genética , Hepatite B/genética , Hepatite B/imunologia , Hepatite C/genética , Hepatite C/imunologia , Humanos , Vacinas de DNA , Vacinas contra Hepatite Viral
18.
Lab Anim (NY) ; 33(7): 37-46, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15224117

RESUMO

Despite the existence of a preventative vaccine, HBV represents a substantial threat to public health, suggesting the need for research to develop new treatments to combat the disease. The authors review the available in vitro and in vivo models, including recently developed transgenic and chimeric mouse models.


Assuntos
Modelos Animais de Doenças , Patos , Vírus da Hepatite B/fisiologia , Hepatite B , Mamíferos , Animais , Técnicas de Cultura de Células/métodos , Camundongos , Camundongos Transgênicos , Fenótipo , Transgenes/genética
19.
Curr Pathobiol Rep ; 2(1): 11-20, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25650171

RESUMO

During the past decade, a series of discoveries has established the potential of the so called terminally differentiated cells to transition to more primitive progenitor cells. The dramatic demonstration of the ability to reprogram differentiated somatic cells to induced pluripotent stem cells (iPSC) that can then give rise to cells of all three germ layers has opened the possibility of generating virtually any cell type in culture, from any given individual. Taking advantage of these concepts, researchers have generated iPSCs by reprogramming a wide variety of somatic cells. In addition to their practical implications, these studies have provided crucial insights into the mechanism of cell plasticity that underlies the transition from one cell type to another. Using concepts derived from research on embryological development, investigators have differentiated iPSCs to cells resembling hepatocytes in many ways. Such hepatocyte-like cells could be of enormous value in disease modeling, drug discovery and regenerative medicine. However, the currently available methods do not yield cells that fully reproduce the characteristics of adult primary hepatocytes. Thus generating hepatocytes from iPSCs is very much a work in progress. In addition to chronicling these exciting developments, this review will discuss the emergent new approaches to generating iPSCs, improving their differentiation to hepatocyte-like cells and maintaining the hepatocyte-like cells in culture for longer survival and better function.

20.
PLoS One ; 7(10): e46775, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23091601

RESUMO

BACKGROUND AND AIMS: Preparative hepatic irradiation (HIR), together with mitotic stimulation of hepatocytes, permits extensive hepatic repopulation by transplanted hepatocytes in rats and mice. However, whole liver HIR is associated with radiation-induced liver disease (RILD), which limits its potential therapeutic application. In clinical experience, restricting HIR to a fraction of the liver reduces the susceptibility to RILD. Here we test the hypothesis that repopulation of selected liver lobes by regional HIR should be sufficient to correct some inherited metabolic disorders. METHODS: Hepatocytes (10(7)) isolated from wildtype F344 rats or Wistar-RHA rats were engrafted into the livers of congeneic dipeptidylpeptidase IV deficient (DPPIV(-)) rats or uridinediphosphoglucuronateglucuronosyltransferase-1A1-deficient jaundiced Gunn rats respectively by intrasplenic injection 24 hr after HIR (50 Gy) targeted to the median lobe, or median plus left liver lobes. An adenovector expressing hepatocyte growth factor (10(11) particles) was injected intravenously 24 hr after transplantation. RESULTS: Three months after hepatocyte transplantation in DPPIV(-) rats, 30-60% of the recipient hepatocytes were replaced by donor cells in the irradiated lobe, but not in the nonirradiated lobes. In Gunn rats receiving median lobe HIR, serum bilirubin declined from pretreatment levels of 5.17 ± 0.78 mg/dl to 0.96 ± 0.30 mg/dl in 8 weeks and remained at this level throughout the 16 week observation period. A similar effect was observed in the group, receiving median plus left lobe irradiation. CONCLUSIONS: As little as 20% repopulation of 30% of the liver volume was sufficient to correct hyperbilirubinemia in Gunn rats, highlighting the potential of regiospecific HIR in hepatocyte transplantation-based therapy of inherited metabolic liver diseases.


Assuntos
Hepatócitos/transplante , Icterícia/terapia , Regeneração Hepática , Animais , Bile/metabolismo , Bilirrubina/sangue , Bilirrubina/metabolismo , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Ativação Enzimática , Glucuronosiltransferase/metabolismo , Hepatócitos/efeitos da radiação , Icterícia/metabolismo , Fígado/metabolismo , Fígado/efeitos da radiação , Fígado/cirurgia , Regeneração Hepática/efeitos da radiação , Masculino , Ratos , Ratos Transgênicos , Condicionamento Pré-Transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA